Play all audios:
ABSTRACT Interferon regulatory factor 4 (IRF4) is an attractive therapeutic target in multiple myeloma (MM). We here report that expression of IRF4 mRNA inversely correlates with microRNA
(miR)-125b in MM patients. Moreover, we provide evidence that miR-125b is downregulated in TC2/3 molecular MM subgroups and in established cell lines. Importantly, constitutive expression of
miR-125b-5p by lentiviral vectors or transfection with synthetic mimics impaired growth and survival of MM cells and overcame the protective role of bone marrow stromal cells _in vitro_.
Apoptotic and autophagy-associated cell death were triggered in MM cells on miR-125b-5p ectopic expression. Importantly, we found that the anti-MM activity of miR-125b-5p was mediated via
direct downregulation of IRF4 and its downstream effector BLIMP-1. Moreover, inhibition of IRF4 translated into downregulation of c-Myc, caspase-10 and cFlip, relevant IRF4-downstream
effectors. Finally, _in vivo_ intra-tumor or systemic delivery of formulated miR-125b-5p mimics against human MM xenografts in severe combined immunodeficient/non-obese diabetic mice induced
significant anti-tumor activity and prolonged survival. Taken together, our findings provide evidence that miR-125b, differently from other hematologic malignancies, has tumor-suppressor
activity in MM. Furthermore, our data provide proof-of-concept that synthetic miR-125b-5p mimics are promising anti-MM agents to be validated in early clinical trials. SIMILAR CONTENT BEING
VIEWED BY OTHERS THERAPEUTIC TARGETING MIR130B COUNTERACTS DIFFUSE LARGE B-CELL LYMPHOMA PROGRESSION VIA OX40/OX40L-MEDIATED INTERACTION WITH TH17 CELLS Article Open access 18 March 2022
ANTI-BACTERIAL AND ANTI-VIRAL NANCHANGMYCIN DISPLAYS ANTI-MYELOMA ACTIVITY BY TARGETING OTUB1 AND C-MAF Article Open access 30 September 2020 AN MTORC1 TO HRI SIGNALING AXIS PROMOTES
CYTOTOXICITY OF PROTEASOME INHIBITORS IN MULTIPLE MYELOMA Article Open access 18 November 2022 INTRODUCTION Multiple myeloma (MM) is a genetically complex malignancy from the outset, with
progressive acquisition of genetic lesions mediating drug resistance and high disease burden.1 Despite recent progress in the understanding MM pathobiology and the availability of innovative
drugs which have improved clinical outcome, the disease eventually progresses to a drug-resistant lethal stage (plasma cell leukemia)2, 3, 4 and novel therapeutic strategies are therefore
eagerly awaited. Indeed, one of the major challenges in treating MM is its genomic and phenotypic heterogeneity.5 Hence, an optimal therapy would target an essential regulatory pathway
shared by all disease subsets.6 Interferon regulatory factor 4 (IRF4) is a lymphocyte-specific transcription factor.7 Interference with IRF4 expression is lethal for MM cells, irrespective
of their genetics, making IRF4 an ‘Achilles’ heel’ that may be exploited therapeutically.8 Specifically, IRF4 is oncogenic and overexpressed when translocated to actively transcribed genomic
regions in some MM patients, but it also has a survival effect in MM cells in the absence of translocations or overexpression.7, 8 A relevant IRF4 target gene is c-Myc,7, 8 which has a
prominent role in the pathogenesis of MM.7, 8 Another downstream IRF4 effector is B-lymphocyte-induced maturation protein-1 (BLIMP-1):9 indeed, knockdown of BLIMP-1 causes apoptosis in MM
cells. These findings suggest that IRF4 may regulate MM cell survival through modulation of BLIMP-1.9 Moreover, it has been recently demonstrated that caspase-10 (casp-10) and cFlip genes
are transactivated by IRF4: importantly, the evidence that all MM cell lines require casp-10 and cFLIP for survival led to the hypothesis that loss of the proteolytic activity of the
casp-10/cFlip heterodimer mediates MM cell death induced by IRF4 knockdown.10 All these data indicate IRF4 as an attractive therapeutic target in MM. However, efficient _in vivo_ strategies
aimed at blocking IRF4 pathway are still lacking. MicroRNAs (miRNAs) are small non-coding RNAs of 19–25 nucleotides, which regulate gene expression by degrading or inhibiting translation of
target mRNAs, primarily via base pairing to partially or fully complementary sites in the 3′ untranslated region (UTR).11 Targeting deregulated miRNAs in cancer cells is emerging as a novel
promising therapeutic approach,12, 13, 14 including in MM.15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34 In this scenario, replacement of tumor-suppressor
miRNAs by synthetic oligonucleotides (miRNA mimics) offers a new therapeutic opportunity to restore a loss-of-function in cancer, that has been an unmet need for drug developers.35 Here, we
show that IRF4 expression is regulated by microRNA-125b-5p (miR-125b-5p) in patient-derived MM cells and MM cell lines. In most of these cells, enforced expression of miR-125b-5p affects
growth and survival, acting via IRF4 downregulation and impairment of its downstream signaling. Overall, our findings demonstrate that miR-125b is a tumor suppressor in MM, and provide the
rationale for development of miR-125b-5p mimics as novel therapeutics. MATERIALS AND METHODS MM PATIENT CELLS AND CELL LINES Following the Magna Graecia University IRB study approval,
primary MM cells were isolated from bone marrow (BM) aspirates, as described,19 from 24 newly diagnosed MM patients who had provided the informed consent. For transfection purposes and
proliferation/survival assays, peripheral blood mononuclear cells (PBMCs) from healthy donors have been used as controls. MM cell lines were cultured as described.19 HS-5 human stromal cell
line (purchased from ATCC, Manassas, VA, USA, CRL-11882) was cultured in Dulbecco's Modified Eagle Medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (see
Supplementary Methods for detailed information). VIRUS GENERATION AND INFECTION OF CELLS Cells stably expressing green fluorescent protein transgene were obtained as described.21 To generate
cells stably expressing luciferase transgene, NCI-H929 cells were transduced with pLenti-III-PGK-Luc (ABM Inc., Richmond, BC, Canada) vector. MM cells stably expressing miR-125b-1 and
miR-125b-2 genes were transduced with Lenti-miR-125b-1 and Lenti-miR-125b-2 miRNA precursor constructs (System Biosciences, CA, USA); lentiviral particles were produced and transduced as
previously described.19 RNA EXTRACTION AND QRT-PCR. RNA samples of healthy donors BM-derived plasma cells were purchased (AllCells LLC, Alameda, CA, USA). Total RNA extraction from MM cells
and quantitative real-time PCR were performed as previously described (see Supplementary Methods for detailed information).19 _IN VITRO_ TRANSFECTION OF MM CELLS Synthetic miRNA mimics were
purchased from Ambion (Applied Biosystems, Carlsbad, CA, USA), while synthetic miRNA inhibitors were purchased from Exiqon (Vedbaek, Rudersdal, Denmark). Silencer Select siRNAs were
purchased from Ambion (Applied Biosystems). All the oligos were used at 100 nmol/l final concentration. A total of 2,5 × 105 cells were transfected using Neon Transfection System
(Invitrogen, Carlsbad, CA, USA) (2 pulse at 1.050 V, 30 ms) and the transfection efficiency, evaluated by flow-cytometric analysis relative to a FAM dye-labeled miRNA inhibitor negative
control, reached 85 to 90%. The same conditions were applied for transfection of MM cells with 5 μg of expression vectors carrying the open reading frames (ORFs) of BLIMP-1 (EX-Z5827-M11),
IRF4 (EX-M0891-M11) or empty vector (EX-EGFP-M11) (GeneCopeia, Rockville, MD, USA). SURVIVAL ASSAY Cell viabilty was evaluated by Cell Counting Kit-8 (CCK-8) assay (Dojindo Molecular
Technologies, Mashikimachi, Japan) and 7-Aminoactinomycin (7-AAD) flow cytometry assay (BD Biosciences, San Jose, CA, USA), according to manufacturer’s instructions. DETECTION OF APOPTOSIS
Apoptosis was investigated by three different assays: Annexin V/7-AAD flow cytometry assay, terminal-deoxynucleotidyl transferase-dUTP nick end labeling (TUNEL) assay and western blot
analysis of caspases expression and cleavage. To perform TUNEL assay, transfected cells were fixed using 4% paraformaldehyde and then permeabilized using 0.25% Triton X-100, according to
manufacturer’s instructions. Cells were stained with a TUNEL assay (Click-iT TUNEL Alexa Fluor 594 Imaging Assay, Invitrogen, 10246) to identify those with fragmented DNA. Nuclei were
counterstained with Hoechst 33342 (Life Technologies, CA, USA). Image acquisition was done using EVOS FLOID (Life Technologies) equipped with a 20 × Nikon objective. WESTERN BLOT ANALYSIS
Whole cell protein extracts were prepared from MM cell lines and from PBMCs in NP40 CellLysis Buffer (Life Technologies) containing a cocktail of protease inhibitors (Sigma, Steinheim,
Germany). Cell lysates were loaded and polyacrylamide gel electrophoresis separated. Proteins were transferred by Trans-Blot Turbo Transfer Starter System (Bio-Rad, Hercules, CA, USA) for 7
min. After protein transfer, the membranes were blotted with the primary antibodies (see Supplementary methods for detailed information). ANIMALS AND _IN VIVO_ MODEL OF HUMAN MM Male CB-17
Severe Combined Immunodeficient (SCID) mice (6–8 weeks old; Harlan Laboratories, Inc., Indianapolis, IN, USA) were housed and monitored in our Animal Research Facility. Experimental
procedures and protocols had been approved by the Magna Graecia University IRB and conducted according to protocols approved by the National Directorate of Veterinary Services (Italy, Rome).
Mice were s.c. inoculated with 5 × 106 NCI-H929 cells and treatment started when palpable tumors became detectable. Sample size (that is, number of animals to be inoculated with MM cells)
was chosen accordingly to our experience.16, 17, 18, 19, 20 Tumor sizes were measured as described,19 and the investigator was blinded to group allocation. Tumor size of luciferase
gene-marked NCI-H929 xenografts was also measured by IVIS Lumina II (PerkinElmer, Waltham, MA, USA). Oligos were NLE-formulated within MaxSuppressor _In Vivo_ LANCEr II (BIIo Scientific,
Austin, TX, USA) to achieve an efficient delivery, as reported.16, 27 STATISTICAL ANALYSIS Each experiment was performed at least three times and values are reported as mean±s.d. Comparisons
between groups were made with student’s _t_-test, while statistical significance of differences among multiple groups was determined by GraphPad software (www.graphpad.com). Graphs were
obtained using Graphpad Prism version 6.0 (GraphPad Software, La Jolla, CA, USA). _P_-value <0.05 was accepted as statistically significant. RESULTS INVERSE CORRELATION BETWEEN IRF4 MRNA
AND MIR-125B IN MM PATIENTS To identify IRF4-targeting miRNAs, we interrogated microRNA Data Integration Portal (mirDIP), applying the _high precision_ quality filter.36 As shown in Table 1,
this analysis disclosed 12 mature miRNAs including miR-125a-5p, miR-125b-5p, miR-128, miR-27a-3p, miR-27b-3p, miR-30a-5p, miR-30b-5p, miR-30c-2, miR-30d-5p, miR-30e-5p, miR-4319 and
miR-513a-5p. We next attempted to correlate the expression of these miRNAs and IRF4 mRNA in our dataset (55 MM and 21 plasma cell leukemia patients)(GSE39925). This integrated approach
revealed a significant inverse correlation between IRF4 mRNA and five precursor-miRNAs (pre-miR-125b-1, pre-miR-125b-2, pre-miR-30b, pre-miR-30c-2 and pre-miR-30d) among all MM and plasma
cell leukemia patients examined (Figures 1a and b and Supplementary Fig. S1A). Importantly, when a second dataset (GSE47552) was interrogated, the inverse correlation with IRF4 mRNA was
confirmed only for pre-miR-125b-1 (Supplementary Fig. S1B), strengthening a possible role of miR-125b-5p as IRF4 negative regulator in MM patients. By qRT-PCR, we then evaluated the
expression of miR-125b-5p in 24 CD138+ primary patient MM (ppMM) cells, 10 MM cell lines and 3 samples of CD138+ BM-derived plasma cells from healthy donors (HD-PCs). A significant
downregulation of miR-125b-5p was found in MM cell lines (Figure 1c, with single values plotted in Supplementary Fig. S2), while the downregulatory trend observed in ppMM cells reached
statistical significance within the TC2 and TC3 subgroups only (Figures 1c and d). ENFORCED EXPRESSION OF MIR-125B IMPAIRS GROWTH AND SURVIVAL OF MM CELLS To evaluate the effects induced by
miR-125b, we transduced 3 MM cell lines (NCI-H929, SK-MM-1, RPMI-8226) with lentiviral vectors carrying either miR-125b-1 or miR-125b-2 (Lenti-miR-125b-1 or Lenti-miR-125b-2) genes. The
effects on cell proliferation were assessed by CCK-8 assay at 2, 3 and 4 days after selection by puromycin. As shown in Figures 2a and b, constitutive expression of either Lenti-miR-125b-1
or Lenti-miR-125b-2 resulted in a strong inhibition of cell growth. Next, we transfected MM cell lines with either synthetic miR-125b-5p mimics or inhibitors. We found that ectopic
expression of miR-125b-5p inhibitors did not affect the proliferation of MM cells (Figures 2c and d); conversely, transfection of miR-125b-5p mimics strongly impaired growth and survival of
most MM cell lines (9 out of 10) (Figures 2e and f). Importantly, miR-125b-5p mimics reduced the viability of ppMM cells from 3 individuals (Figure 2g), but not of PBMCs from 6 healthy
donors (Figure 2h). Taken together, these results indicate that enforced expression of miR-125b-5p inhibits growth and survival of MM cells, consistent with a tumor-suppressor function of
this miRNA. Notably, baseline expression of miR-125b-5p did not correlate with the sensitivity/response to synthetic mimics or inhibitors, suggesting that miR-125b-5p expression is not
predictive of _in vitro_ anti-MM activity. Furthermore, miR-125b-5p mimics were lethal to MM cells irrespective of their genetics. MIR-125B-5P MIMICS INHIBIT PROLIFERATION OF MM CELLS VIA
TARGETING IRF4 To investigate whether IRF4 expression could be affected by ectopic miR-125b-5p, both qRT-PCR and western blot analysis were performed in 3 MM cell lines transfected with
miR-125b-5p mimics or scrambled controls (miR-NC). Specifically, the IRF4 translocated SK-MM-1, along with NCI-H929 and RPMI-8226 cells (not IRF4 translocated), were selected for this
analysis. As shown in Figures 3a and b, transfection of miR-125b-5p downregulated IRF4 at both mRNA and protein levels in all MM cell lines. Notably, the only MM cell line resistant to
miR-125b-5p overexpression lacked detectable IRF4 (i.e. RPMI-8226/Dox40 cells, which were generated from the parental RPMI-8226 by continuous exposure to increasing amounts of doxorubicin to
culture medium, is characterized by loss of miR-125b-5p relevant targets) (Supplementary Fig. S3A). Consistently, we found that viability of RPMI-8226/Dox40 cells was not affected by IRF4
siRNA silencing (Supplementary Fig. S3B); in contrast, siRNA-transfection of both NCI-H929 and SK-MM-1 cells confirmed its role in supporting their survival (Supplementary Fig. S3B). We next
investigated whether ectopic expression of a cDNA containing only the coding region of IRF4 and lacking the miR-125b-5p-targeted 3′ UTR could protect MM cells from miR-125b-5p
anti-proliferative effects. As shown in Figure 3c, transfection of IRF4 construct increased IRF4 protein expression which was not affected by miR-125b-5p mimics. Importantly, IRF4
overexpression rescued SK-MM-1 cells from the growth-inhibitory activity of either IRF4 siRNAs (Supplementary Fig. S3C) or miR-125b-5p mimics (Figure 3d), indicating that miR-125b-5p exerts
its anti-MM activity via targeting IRF4. ENFORCED EXPRESSION OF MIR-125B-5P IMPAIRS IRF4 SIGNALING IN MM CELL LINES We next investigated the effects of miR-125b-5p on the molecular network
underlying IRF4 activity in MM. Interestingly, the IRF4-downstream effector BLIMP-1 has been proven to be a direct target of miR-125b-5p.37 As shown in Figure 4a and Supplementary Fig. S4A,
transfection of miR-125b-5p mimics downregulated BLIMP-1 protein in SK-MM-1 and NCI-H929 cells. Thus, we investigated whether ectopic expression of a cDNA containing only the coding region
of BLIMP-1 and lacking the miR-125b-5p-targeted 3′ UTR could protect MM cells from miR-125b-5p effects. Importantly, co-transfection with BLIMP-1 construct weakened the anti-proliferative
effect of miR-125b-5p mimics as well of BLIMP-1 siRNAs (Figure 4c and Supplementary Fig. S4B), indicating that also BLIMP-1 mediates the anti-MM activity of miR-125b-5p. Of note,
miR-125b-5p-induced downregulation of BLIMP-1 was not abrogated in SK-MM-1 cells co-transfected with the coding region of IRF4 (Figure 4b), consistent with the notion that BLIMP-1 is a
direct target of miR-125b-5p which circumvents IRF4 activity on BLIMP-1. Other IRF4-downstream effectors, with a prominent role in MM pathogenesis, are c-Myc, casp-10 and cFLIP.8, 10 By
western blot analysis, we found reduced expression of these proteins in SK-MM-1 (Figure 4d) and NCI-H929 (Supplementary Fig. S4E) cells at 24–48 h after transfection with miR-125b-5p.
Expression of both casp-10 and cFlip was also reduced at mRNA levels (Supplementary Fig. S4F) by miR-125b-5p mimics. _In silico_ search for target prediction identified both casp-10 and
cFlip as _bona fide_ direct targets for miR-125b-5p. To validate this interaction in MM cells, SK-MM-1 cells were co-transfected with miR-125b-5p mimics or scrambled oligonucleotides,
together with an expression vector carrying the 3′ UTR of casp-10 or cFlip mRNA cloned downstream of luciferase reporter gene. Of note, we did not find significant changes in 3′ UTR
luciferase activity after miR-125b-5p overexpression, ruling out direct targeting of casp-10 and cFlip mRNAs by miR-125b-5p (Supplementary Fig. S4G). Moreover, miR-125b-5p-induced
downregulation of c-Myc, casp-10 and cFlip was abrogated in SK-MM-1 cells co-transfected with the coding region of IRF4 (Figure 4e), indicating that miR-125b-5p-induced downregulation of
casp-10, c-Myc and cFlip occurs via IRF4 inhibition. Taken together, our findings demonstrate that miR-125b-5p mimics impair IRF4 signaling in MM cells (Figure 4f). We also found that direct
targeting of IRF4 or BLIMP-1 and indirect modulation of c-Myc, casp-10 and cFlip mediates anti-MM activity of this miRNA. MIR-125B-5P MIMICS TRIGGER BOTH APOPTOTIC AND AUTOPHAGY-ASSOCIATED
CELL DEATH Inhibition of IRF4, as well of BLIMP-1 or c-Myc, has been mainly related to induction of apoptosis,9, 38 while targeting casp-10 or its interaction with cFlip triggers autophagic
cell death of MM cells.10 On this basis, we next determined whether apoptotic or autophagy-associated cell death occurred in MM cells with enforced expression of miR-125b-5p. Using Annexin
V/7-AAD flow cytometry assay, we found that miR-125b-5p mimics triggered exposure of phosphatidylserine (PS) and phosphatidylethanolamine (PE) on the cell surface of SK-MM-1 (Figure 5a),
NCI-H929 and U266 cells (Supplementary Fig. S5A), followed by cell membrane disruption. Moreover, by TUNEL assay and western blotting, we found that miR-125b-5p induced DNA-fragmentation
(Supplementary Fig. S5B) and cleavage/activation of both initiator casp-8 and effector casp-3 (Figure 5b and Supplementary Fig. S5C) in MM cells. Of note, apoptosis was not detected when
miR-125b-5p-transfected cells were treated with the pan-caspase inhibitor Z-VAD-fmk (Figure 5c). Overall, these results indicate that miR-125b-5p is pro-apoptotic in MM cells. Moreover, by
flow cytometry analysis of Cyto-ID stained cells, we observed an increase of autophagic vacuoles in SK-MM-1, XG-1 and INA-6 cells, at 48 h after transfection with miR-125b-5p (Figure 5d).
Importantly, the increase of autophagic vacuoles occurred to a similar extent in cells transfected with miR-125b-5p compared with cells starved for 48 h. Moreover, western blot analysis
showed decreased p-62/SQSTM1 and increased Beclin-1 and proteolytic active LCIIIB (Figure 5e), further indicating that miR-125b-5p-induced cell death can be associated with autophagy
induction in MM cells. Indeed, exposure of miR-125b-5p-transfected cells to autophagy inhibitor cloroquine resulted in increased cell death (Figure 5f), thus suggesting a protective role of
autophagy in our experimental settings. MIR-125B-5P MIMICS ANTAGONIZE THE BMSCS PROTECTIVE ROLE ON MM CELLS BM _milieu_ strongly supports survival and proliferation of MM cells.3 In this
regard, we found that exogenous interleukin-6 (IL-6) or insulin-like growth-factor-1 (IGF-1) or hepatocyte growth-factor (HGF) significantly reduced miR-125b-5p expression in all MM cell
lines except U266 cells (Figure 6a), which express L-Myc instead of c-Myc. Since c-Myc suppresses miR-125b transcription39, 40 and is upregulated by IL-6, IGF-1 and HGF,40, 41, 42 we
investigated whether these factors downregulate miR-125b in MM cells through c-Myc induction. Specifically, we treated c-Myc-expressing SK-MM-1 and c-Myc-defective U266 cells with the
10058-F4 small molecule inhibitor of Myc–Max heterodimerization38 and with the JQ1 BET-bromodomain inhibitor, which is reported to inhibit c-Myc transcription.43 Importantly, both compounds
triggered a significant miR-125b-5p upregulation in SK-MM-1 cells, but not in U226 cells (Figure 6b), indicating that c-Myc-independent downregulation of miR-125b occurs in this cell line.
Moreover, IL-6 or IGF-1 or HGF did not affect miR-125b-5p expression in SK-MM-1 cells in the presence of c-Myc inhibitors, indicating a c-Myc-mediated downregulation of miR-125b by exogenous
addition of microenvironmental growth factors (Figure 6c). Finally, we demonstrated that the anti-MM activity of miR-125b-5p mimics was not antagonized by exogenous growth
promoting/pro-survival stimuli including IL-6, IGF-1 and HGF, or by adherence of MM cell lines to HS-5 human stromal cells (Figures 6d and e). This could be partly due to downregulation of
IL-6R/CD126 on the surface of MM cells. Indeed, by flow cytometry analysis, we did observe decreased expression of cell surface IL-6R/CD126, also a further validated target of miR-125b-5p44
(Figure 6f), which translated in reduced levels of phosphorylated/active STAT3 (pSTAT3-Y705) in IL-6-dependent INA-6 cells (Figure 6g). _IN VIVO_ DELIVERY OF NLE-FORMULATED SYNTHETIC
MIR-125B-5P MIMICS EXERTS ANTI-MM ACTIVITY _In vivo_ anti-MM activity of miR-125b-5p was next evaluated in Non-Obese Diabetic (NOD)/SCID mice bearing subcutaneous NCI-H929 xenografts. In our
first model, luciferase gene-marked NCI-H929 xenografts were intra-tumorally treated every other day with 1 mg/kg of oligos for a total of six injections. As shown in Figures 7a–c,
treatment resulted in a significant tumor-growth inhibition and prolonged survival. In a second model, treatments were administered by i.p. injections of formulated oligos (twice weekly, 1
mg/kg). In this model, we also observed anti-tumor activity of miR-125b-5p, evidenced by growth inhibition and prolonged survival (Figures 7d and e). Importantly, we found increased
miR-125b-5p in tumors retrieved from animals 48 h after treatment, confirming efficient tumor cells uptake of formulated oligos (Figure 7f). Consistent with our _in vitro_ data, IRF4
signaling was correlated with miR-125b-5p activity _in vivo_: downregulation of IRF4 and BLIMP-1 (Figure 7g), along with decreased expression of c-Myc, casp-10 and cFlip proteins (Figure
7h), was detected in tumors retrieved from miR-125b-5p-treated animals. Overall, these data indicate that _in vivo_ anti-MM activity of miR-125b-5p mimics is associated with abrogation of
IRF4 signaling within MM xenografts. DISCUSSION In this study, we investigated the anti-MM activity of the IRF4-targeting miR-125b-5p. IRF4 is in fact an ‘Achilles’ heel’ for MM cells and,
therefore, represents an attractive therapeutic target in this malignancy.7 miRNAs are natural antisense interactors of mRNAs, and the availability of suitable _in vivo_ delivery systems has
recently allowed the development of synthetic miRNA mimics in early clinical trials. By querying mirDIP36 and applying the high precision quality filter, we found 12 mature miRNAs predicted
to target the 3′ UTR of IRF4 mRNA, including miR-125b-5p. Importantly, integrated analysis of miRNAs and mRNAs expression profiles showed that miR-125b, but not the other predicted miRNAs,
inversely correlated with IRF4 mRNA in two different MM datasets, strengthening the relevance of miR-125b as IRF4 negative regulator in MM patients. Furthermore, miR-125b-5p was found
significantly downregulated in patients belonging to TC2 and TC3 molecular subgroups, as well as in MM cell lines. Altogether, these findings prompted us to investigate the role of this
miRNA in MM. miR-125b is one of the most evolutionary conserved miRNAs.45 In humans there are two paralogs (hsa-miR-125b-1 on chromosome 11 and hsa-miR-125b-2 on chromosome 21), coding for
the same mature sequences (miR-125b-5p and miR-125b-3p).45 miR-125b has a crucial role in a variety of cellular processes and diseases.46 It is commonly dysregulated in cancer,46 but its
function diverges in different malignancies, with dependence on the molecular contexts. However, its role in MM is still largely undisclosed.46, 47, 48 We here provide the evidence that
miR-125b acts as a tumor suppressor in MM by targeting IRF4 and BLIMP-1 mRNAs, and importantly we show that NLE-formulated synthetic miR-125b-5p mimics induce anti-tumor activity _in vivo_.
Specifically, we demonstrate that both lentivirus-based constitutive expression of miR-125b-1/-2 genes and transient enforced expression of synthetic miR-125b-5p mimics inhibit the growth
and survival of MM cell lines. Moreover, viability of ppMM cells, but not of PBMCs from healthy donors, was affected by transfection with miR-125b-5p mimics, suggesting a favorable
therapeutic activity. In our study, baseline expression of miR-125b-5p does not correlate with _in vitro_ sensitivity of MM cells to synthetic mimics; consistent with our findings,
increasing evidence demonstrate that cancer cells with normal miRNA expression are indeed susceptible to miRNA treatment.35 Importantly, the anti-MM activity of miR-125b-5p mimics was not
affected by either exogenous growth promoting/pro-survival stimuli including IL-6, IGF-1 or HGF, or by adherence of MM cell lines to BM stromal cells (BMSCs). This is a crucial point taking
into account that the close and dynamic interplay between MM cells and BMSCs leads to activation of signal transduction pathways which promote cell-cycle progression and protection from
apoptosis.3 Furthermore, a c-Myc-mediated downregulation of miR-125b by exposure of MM cells to different growth factors was observed. To date, a variety of oncogenic pathways have been
identified as directly regulated by miR-125b.46, 47 Here, we demonstrate a functional link between this miRNA and the oncogenic IRF4 signaling in MM. IRF4 is a validated target of
miR-125b-5p37, 49 and the relevance of this interaction is well established in both myeloid- and B-cell leukemias, wherein IRF4 acts as a tumor suppressor and miR-125b as a tumor promoter.50
We found that IRF4 expression was downregulated on transfection of MM cells with miR-125b-5p mimics. Among the MM cell lines studied, only miR-125b-5p-resistant RPMI-8226/Dox40 cells did
not express detectable levels of IRF4. Importantly, overexpression of IRF4 was able to rescue SK-MM-1 cells from the growth-inhibitory activity of miR-125b-5p. Altogether, these data
indicate that IRF4 mediates the anti-MM activity of miR-125b-5p mimics _in vitro_, underlying a novel and divergent role of miR-125b-5p/IRF4 axis in MM as compared with other hematological
malignancies. miRNAs function as master regulators of the genome by modulating the expression of tens to hundreds genes, often belonging to the same pathway.35 This mechanism of action
provides advantage to the therapeutic use of miRNAs, since it is consistent with the current vision of cancer as a pathway disease.35 Interestingly, the IRF4-downstream effector BLIMP-1 is
also regulated by miR-125b-5p,37 and we demonstrated that, similar to IRF4 but to a lesser extent, it also mediates the growth-inhibitory activity of miR-125b-5p. Moreover, we analyzed
perturbations occurring in other relevant effectors of IRF4 signaling, including c-Myc, casp-10 and cFlip. c-Myc has a prominent role in the pathogenesis of MM, wherein it represents an
attractive therapeutic target.7, 38, 43 Importantly, ectopic expression of miR-125b-5p significantly decreased c-Myc protein in MM cells which was rescued by co-transfection with the coding
region of IRF4, indicating that IRF4 mediates miR-125b-5p-triggered downregulation of c-Myc. Recently, it has been demonstrated that MM cells further require the IRF4-downstream effectors
casp-10 and cFlip for their survival: indeed, the heterodimeric protease composed of casp-10 and cFLIP proteins has a balancing role among the pro-survival and pro-death effects of
autophagy.10 Consistent with these notions, we found that both casp-10 and cFlip undergo significant downregulation on miR-125b-5p ectopic expression in MM cells, along with increased
autophagic flux. Nonetheless, the role of autophagy as a response to miR-125b-5p overexpression points to further investigation, taking into account that our present data indeed suggest a
protective effect at least in SK-MM-1 cells. Even though _in silico_ search for target prediction indicated both casp-10 and cFlip as _bona fide_ direct targets for miR-125b-5p, we failed to
validate this interaction. However, miR-125b-5p-induced downregulation of casp-10 and cFlip was abrogated by ectopic IRF4 expression, indicating that miR-125b-5p-mediated downregulation of
these two survival factors depends on IRF4 targeting. Finally, we demonstrated the _in vivo_ anti-tumor activity of NLE-formulated miR-125b-5p mimics against human MM xenografts in SCID/NOD
mice. To our knowledge, this is the first evidence of a successful _in vivo_ treatment with miR-125b-5p mimics in a murine xenograft model of human MM, which indeed has important potential
towards clinical applications. We showed that both intra-tumor and i.p. injection of NLE-formulated miR-125b-5p mimics resulted in significant tumor-growth inhibition and prolonged survival.
Moreover, in tumors retrieved from animals treated with miR-125b-5p mimics, a downregulation of its direct targets IRF4 and BLIMP-1, along with a reduction in expression of c-Myc, casp-10
and cFlip proteins, was observed. These findings suggest that the _in vivo_ anti-MM activity of miR-125b-5p mimics is related to the impairment of IRF4 signaling within MM xenografts.
Further _in vivo_ evaluation in preclinical models recapitulating the huBMM, such as the SCID-synth-hu,51 will strengthen the translational value of miR-125b-5p mimics. We think that our
study provides important proof-of-concept findings for the basic strategy of miRNA therapeutics. In fact, we found that the miR-125b-5p/IRF4 axis has a highly disease-specific functional
role in MM, which runs in opposite directions as compared with other hematological malignancies. These findings support the peculiarity of MM BM microenvironment disease scenario, which
opens highly specific therapeutic avenues. An additional important point is that miR-125b-5p strongly inhibits both IRF4 and BLIMP-1, offering a pathway-directed therapeutic tool, which is
still undruggable by alternative approaches. In conclusion, our investigation provides evidence that miR-125b has tumor-suppressor activity in MM and that enforced expression of synthetic
miR-125b-5p mimics induces significant anti-MM activity _in vitro and in vivo_ by IRF4 targeting. Taken together, these results provide the rational framework for development of
miR-125b-5p-based therapies in MM. REFERENCES * Anderson KC, Carrasco RD . Pathogenesis of myeloma. _Annu Rev Pathol_ 2011; 6: 249–274. Article CAS PubMed Google Scholar * Rajkumar SV .
Treatment of multiple myeloma. _Nat Rev Clin Oncol_ 2011; 8: 479–491. Article CAS PubMed PubMed Central Google Scholar * Podar K, Chauhan D, Anderson KC . Bone marrow microenvironment
and the identification of new targets for myeloma therapy. _Leukemia_ 2009; 23: 10–24. Article CAS PubMed Google Scholar * Rossi M, Di Martino MT, Morelli E, Leotta M, Rizzo A, Grimaldi
A _et al_. Molecular targets for the treatment of multiple myeloma. _Curr Cancer Drug Targets_ 2012; 12: 757–767. Article CAS PubMed Google Scholar * Morgan GJ, Walker BA, Davies FE .
The genetic architecture of multiple myeloma. _Nat Rev Cancer_ 2012; 12: 335–348. Article CAS PubMed Google Scholar * Kuehl WM, Bergsagel PL . Molecular pathogenesis of multiple myeloma
and its premalignant precursor. _J Clin Invest_ 2012; 122: 3456–3463. Article CAS PubMed PubMed Central Google Scholar * Shaffer AL, Emre NC, Romesser PB, Staudt LM . IRF4: immunity.
Malignancy! Therapy? _Clin Cancer Res_ 2009; 15: 2954–2961. Article CAS PubMed PubMed Central Google Scholar * Shaffer AL, Emre NC, Lamy L, Ngo VN, Wright G, Xiao W _et al_. IRF4
addiction in multiple myeloma. _Nature_ 2008; 454: 226–231. Article CAS PubMed PubMed Central Google Scholar * Lin FR, Kuo HK, Ying HY, Yang FH, Lin KI . Induction of apoptosis in
plasma cells by B lymphocyte-induced maturation protein-1 knockdown. _Cancer Res_ 2007; 67: 11914–11923. Article CAS PubMed Google Scholar * Lamy L, Ngo VN, Emre NC, Shaffer AL 3rd, Yang
Y, Tian E _et al_. Control of autophagic cell death by caspase-10 in multiple myeloma. _Cancer Cell_ 2013; 23: 435–449. Article CAS PubMed PubMed Central Google Scholar * Bartel DP .
MicroRNAs: genomics, biogenesis, mechanism, and function. _Cell_ 2004; 116: 281–297. Article CAS PubMed Google Scholar * Garzon R, Marcucci G, Croce CM . Targeting microRNAs in cancer:
rationale, strategies and challenges. _Nat Rev Drug Discov_ 2010; 9: 775–789. Article CAS PubMed PubMed Central Google Scholar * Trang P, Weidhaas JB, Slack FJ . MicroRNAs as potential
cancer therapeutics. _Oncogene_ 2008; 27: S52–S57. Article CAS PubMed Google Scholar * Misso G, Di Martino MT, De Rosa G, Farooqi AA, Lombardi A, Campani V _et al_. Mir-34: a new weapon
against cancer? _Mol Ther Nucleic Acids_ 2014; 3: e194. Article CAS PubMed Google Scholar * Amodio N, Di Martino MT, Neri A, Tagliaferri P, Tassone P . Non-coding RNA: a novel
opportunity for the personalized treatment of multiple myeloma. _Exp Opin Biol Ther_ 2013; 13: S125–S137. Article CAS Google Scholar * Di Martino MT, Leone E, Amodio N, Foresta U,
Lionetti M, Pitari MR _et al_. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: _in vitro_ and _in vivo_ evidence. _Clin Cancer Res_ 2012; 18: 6260–6270. Article
CAS PubMed PubMed Central Google Scholar * Di Martino MT, Gulla A, Gallo Cantafio ME, Altomare E, Amodio N, Leone E _et al_. _In vitro_ and _in vivo_ activity of a novel locked nucleic
acid (LNA)-inhibitor-miR-221 against multiple myeloma cells. _PloS One_ 2014; 9: e89659. Article PubMed PubMed Central Google Scholar * Di Martino MT, Gulla A, Cantafio ME, Lionetti M,
Leone E, Amodio N _et al_. _In vitro_ and _in vivo_ anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. _Oncotarget_ 2013; 4: 242–255. Article PubMed PubMed Central Google
Scholar * Leone E, Morelli E, Di Martino MT, Amodio N, Foresta U, Gulla A _et al_. Targeting miR-21 inhibits _in vitro_ and _in vivo_ multiple myeloma cell growth. _Clin Cancer Res_ 2013;
19: 2096–2106. Article CAS PubMed PubMed Central Google Scholar * Amodio N, Di Martino MT, Foresta U, Leone E, Lionetti M, Leotta M _et al_. miR-29b sensitizes multiple myeloma cells to
bortezomib-induced apoptosis through the activation of a feedback loop with the transcription factor Sp1. _Cell Death Dis_ 2012; 3: e436. Article CAS PubMed PubMed Central Google
Scholar * Leotta M, Biamonte L, Raimondi L, Ronchetti D, Di Martino MT, Botta C _et al_. A p53-dependent tumor suppressor network is induced by selective miR-125a-5p inhibition in multiple
myeloma cells. _J Cell Physiol_ 2014; 229: 2106–2116. Article CAS PubMed Google Scholar * Zhao JJ, Lin J, Zhu D, Wang X, Brooks D, Chen M _et al_. miR-30-5p functions as a tumor
suppressor and novel therapeutic tool by targeting the oncogenic Wnt/beta-catenin/BCL9 pathway. _Cancer Res_ 2014; 74: 1801–1813. Article CAS PubMed PubMed Central Google Scholar *
Misiewicz-Krzeminska I, Sarasquete ME, Quwaider D, Krzeminski P, Ticona FV, Paino T _et al_. Restoration of microRNA-214 expression reduces growth of myeloma cells through positive
regulation of P53 and inhibition of DNA replication. _Haematologica_ 2013; 98: 640–648. Article CAS PubMed PubMed Central Google Scholar * Pichiorri F, Suh SS, Ladetto M, Kuehl M,
Palumbo T, Drandi D _et al_. MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. _Proc Natl Acad Sci USA_ 2008; 105: 12885–12890. Article CAS PubMed PubMed
Central Google Scholar * Pichiorri F, Suh SS, Rocci A, De Luca L, Taccioli C, Santhanam R _et al_. Downregulation of p53-inducible microRNAs 192, 194, and 215 impairs the p53/MDM2
autoregulatory loop in multiple myeloma development. _Cancer Cell_ 2010; 18: 367–381. Article CAS PubMed PubMed Central Google Scholar * Rossi M, Amodio N, Di Martino MT, Tagliaferri P,
Tassone P, Cho WC . MicroRNA and multiple myeloma: from laboratory findings to translational therapeutic approaches. _Curr Pharma Biotechnol_ 2014; 15: 459–467. Article CAS Google Scholar
* Raimondi L, Amodio N, Di Martino MT, Altomare E, Leotta M, Caracciolo D _et al_. Targeting of multiple myeloma-related angiogenesis by miR-199a-5p mimics: _in vitro_ and _in vivo_
anti-tumor activity. _Oncotarget_ 2014; 5: 3039–3054. Article PubMed PubMed Central Google Scholar * Scognamiglio I, Di Martino MT, Campani V, Virgilio A, Galeone A, Gulla A _et al_.
Transferrin-conjugated SNALPs encapsulating 2'-O-methylated miR-34a for the treatment of multiple myeloma. _BioMed Res Int_ 2014; 2014: 217365. Article PubMed PubMed Central Google
Scholar * Di Martino MT, Campani V, Misso G, Gallo Cantafio ME, Gulla A, Foresta U _et al_. _In vivo_ activity of miR-34a mimics delivered by stable nucleic acid lipid particles (SNALPs)
against multiple myeloma. _PloS One_ 2014; 9: e90005. Article PubMed PubMed Central Google Scholar * Amodio N, Bellizzi D, Leotta M, Raimondi L, Biamonte L, D'Aquila P _et al_.
miR-29b induces SOCS-1 expression by promoter demethylation and negatively regulates migration of multiple myeloma and endothelial cells. _Cell Cycle_ 2013; 12: 3650–3662. Article CAS
PubMed PubMed Central Google Scholar * Lionetti M, Musto P, Di Martino MT, Fabris S, Agnelli L, Todoerti K _et al_. Biological and clinical relevance of miRNA expression signatures in
primary plasma cell leukemia. _Clin Cancer Res_ 2013; 19: 3130–3142. Article CAS PubMed Google Scholar * Rossi M, Pitari MR, Amodio N, Di Martino MT, Conforti F, Leone E _et al_. miR-29b
negatively regulates human osteoclastic cell differentiation and function: implications for the treatment of multiple myeloma-related bone disease. _J Cell Physiol_ 2013; 228: 1506–1515.
Article CAS PubMed Google Scholar * Amodio N, Leotta M, Bellizzi D, Di Martino MT, D'Aquila P, Lionetti M _et al_. DNA-demethylating and anti-tumor activity of synthetic miR-29b
mimics in multiple myeloma. _Oncotarget_ 2012; 3: 1246–1258. Article PubMed PubMed Central Google Scholar * Tagliaferri P, Rossi M, Di Martino MT, Amodio N, Leone E, Gulla A _et al_.
Promises and challenges of MicroRNA-based treatment of multiple myeloma. _Curr Cancer Drug Targets_ 2012; 12: 838–846. Article CAS PubMed PubMed Central Google Scholar * Bader AG .
miR-34 - a microRNA replacement therapy is headed to the clinic. _Front Genet_ 2012; 3: 120. Article CAS PubMed PubMed Central Google Scholar * Shirdel EA, Xie W, Mak TW, Jurisica I .
NAViGaTing the micronome—using multiple microRNA prediction databases to identify signalling pathway-associated microRNAs. _PloS One_ 2011; 6: e17429. Article CAS PubMed PubMed Central
Google Scholar * Gururajan M, Haga CL, Das S, Leu CM, Hodson D, Josson S _et al_. MicroRNA 125b inhibition of B cell differentiation in germinal centers. _Int Immunol_ 2010; 22: 583–592.
Article CAS PubMed PubMed Central Google Scholar * Holien T, Vatsveen TK, Hella H, Waage A, Sundan A . Addiction to c-MYC in multiple myeloma. _Blood_ 2012; 120: 2450–2453. Article CAS
PubMed Google Scholar * Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM _et al_. Widespread microRNA repression by Myc contributes to tumorigenesis. _Nat Genet_ 2008; 40: 43–50.
Article CAS PubMed Google Scholar * Popowski M, Ferguson HA, Sion AM, Koller E, Knudsen E, Van Den Berg CL . Stress and IGF-I differentially control cell fate through mammalian target of
rapamycin (mTOR) and retinoblastoma protein (pRB). _J Biol Chem_ 2008; 283: 28265–28273. Article CAS PubMed PubMed Central Google Scholar * Shi Y, Frost P, Hoang B, Benavides A, Gera
J, Lichtenstein A . IL-6-induced enhancement of c-Myc translation in multiple myeloma cells: critical role of cytoplasmic localization of the rna-binding protein hnRNP A1. _J Biol Chem_
2011; 286: 67–78. Article CAS PubMed Google Scholar * Li X, Bian Y, Takizawa Y, Hashimoto T, Ikoma T, Tanaka J _et al_. ERK-dependent downregulation of Skp2 reduces Myc activity with
HGF, leading to inhibition of cell proliferation through a decrease in Id1 expression. _Mol Cancer Res_ 2013; 11: 1437–1447. Article CAS PubMed Google Scholar * Delmore JE, Issa GC,
Lemieux ME, Rahl PB, Shi J, Jacobs HM _et al_. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. _Cell_ 2011; 146: 904–917. Article CAS PubMed PubMed Central Google
Scholar * Gong J, Zhang JP, Li B, Zeng C, You K, Chen MX _et al_. MicroRNA-125b promotes apoptosis by regulating the expression of Mcl-1, Bcl-w and IL-6R. _Oncogene_ 2013; 32: 3071–3079.
Article CAS PubMed Google Scholar * Shaham L, Binder V, Gefen N, Borkhardt A, Izraeli S . MiR-125 in normal and malignant hematopoiesis. _Leukemia_ 2012; 26: 2011–2018. Article CAS
PubMed Google Scholar * Sun YM, Lin KY, Chen YQ . Diverse functions of miR-125 family in different cell contexts. _J Hematol Oncol_ 2013; 6: 6. Article CAS PubMed PubMed Central Google
Scholar * Kumar M, Lu Z, Takwi AA, Chen W, Callander NS, Ramos KS _et al_. Negative regulation of the tumor suppressor p53 gene by microRNAs. _Oncogene_ 2011; 30: 843–853. Article CAS
PubMed Google Scholar * Murray MY, Rushworth SA, Zaitseva L, Bowles KM, Macewan DJ . Attenuation of dexamethasone-induced cell death in multiple myeloma is mediated by miR-125b expression.
_Cell Cycle_ 2013; 12: 2144–2153. Article CAS PubMed PubMed Central Google Scholar * Chaudhuri AA, So AY, Sinha N, Gibson WS, Taganov KD, O'Connell RM _et al_. MicroRNA-125b
potentiates macrophage activation. _J Immunol_ 2011; 187: 5062–5068. Article CAS PubMed Google Scholar * So AY, Sookram R, Chaudhuri AA, Minisandram A, Cheng D, Xie C _et al_. Dual
mechanisms by which miR-125b represses IRF4 to induce myeloid and B-cell leukemias. _Blood_ 2014; 124: 1502–1512. Article CAS PubMed PubMed Central Google Scholar * Calimeri T, Battista
E, Conforti F, Neri P, Di Martino MT, Rossi M _et al_. A unique three-dimensional SCID-polymeric scaffold (SCID-synth-hu) model for _in vivo_ expansion of human primary multiple myeloma
cells. _Leukemia_ 2011; 25: 707–711. Article CAS PubMed PubMed Central Google Scholar Download references ACKNOWLEDGEMENTS This work has been supported by funds of Italian Association
for Cancer Research (AIRC), PI: PT. ‘Special Program Molecular Clinical Oncology—5 per mille’ n. 9980, 2010/15. This work has also been supported by a grant from NIH PO1-155258, RO1-124929,
P50-100007, PO1-78378 and VA merit grant IO1-24467. Nicola Amodio was supported by a ‘Fondazione Umberto Veronesi’ post-doctoral fellowship. AUTHOR CONTRIBUTIONS EM, EL, MEGC, MTDM, NA, LB,
AG, UF, MRP, CB and MR performed experiments and analyzed the data; AN provided biological samples; PT and PT conceived the study; EM, PT and PT wrote the manuscript; NCM and KCA provided
critical evaluation of experimental data and manuscript. AUTHOR INFORMATION AUTHORS AND AFFILIATIONS * Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore
Venuta University Campus, Catanzaro, Italy E Morelli, E Leone, M E Gallo Cantafio, M T Di Martino, N Amodio, L Biamonte, A Gullà, U Foresta, M R Pitari, C Botta, M Rossi, P Tagliaferri &
P Tassone * Department of Medical Sciences, University of Milan, Hematology 1, IRCCS Policlinico Foundation, Milan, Italy A Neri * Department of Medical Oncology, Jerome Lipper Multiple
Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA N C Munshi & K C Anderson * VA Boston Healthcare System, West Roxbury, Boston, MA, USA N C Munshi * Sbarro Institute for
Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA P Tassone Authors * E Morelli View author
publications You can also search for this author inPubMed Google Scholar * E Leone View author publications You can also search for this author inPubMed Google Scholar * M E Gallo Cantafio
View author publications You can also search for this author inPubMed Google Scholar * M T Di Martino View author publications You can also search for this author inPubMed Google Scholar * N
Amodio View author publications You can also search for this author inPubMed Google Scholar * L Biamonte View author publications You can also search for this author inPubMed Google Scholar
* A Gullà View author publications You can also search for this author inPubMed Google Scholar * U Foresta View author publications You can also search for this author inPubMed Google
Scholar * M R Pitari View author publications You can also search for this author inPubMed Google Scholar * C Botta View author publications You can also search for this author inPubMed
Google Scholar * M Rossi View author publications You can also search for this author inPubMed Google Scholar * A Neri View author publications You can also search for this author inPubMed
Google Scholar * N C Munshi View author publications You can also search for this author inPubMed Google Scholar * K C Anderson View author publications You can also search for this author
inPubMed Google Scholar * P Tagliaferri View author publications You can also search for this author inPubMed Google Scholar * P Tassone View author publications You can also search for this
author inPubMed Google Scholar CORRESPONDING AUTHOR Correspondence to P Tassone. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no conflict of interest. ADDITIONAL INFORMATION
Supplementary Information accompanies this paper on the Leukemia website SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION (DOC 1888 KB) SUPPLEMENTARY FIGURES (DOC 33 KB) RIGHTS AND
PERMISSIONS This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License. The images or other third party material in this article are included
in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain
permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/ Reprints and permissions ABOUT THIS
ARTICLE CITE THIS ARTICLE Morelli, E., Leone, E., Cantafio, M. _et al._ Selective targeting of IRF4 by synthetic microRNA-125b-5p mimics induces anti-multiple myeloma activity _in vitro_ and
_in vivo_. _Leukemia_ 29, 2173–2183 (2015). https://doi.org/10.1038/leu.2015.124 Download citation * Received: 19 February 2015 * Revised: 27 April 2015 * Accepted: 05 May 2015 * Published:
19 May 2015 * Issue Date: November 2015 * DOI: https://doi.org/10.1038/leu.2015.124 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get
shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative